PD1 Expression in EGFRvIII-Directed CAR T Cell Infusion Product for Glioblastoma Is Associated with Clinical Response

Oliver Y Tang, Lifeng Tian, Todd Yoder, Rong Xu, Irina Kulikovskaya, Minnal Gupta, Jan Joseph Melenhorst, Simon F Lacey, Donald M O'Rourke, Zev A Binder, Oliver Y Tang, Lifeng Tian, Todd Yoder, Rong Xu, Irina Kulikovskaya, Minnal Gupta, Jan Joseph Melenhorst, Simon F Lacey, Donald M O'Rourke, Zev A Binder

Abstract

The epidermal growth factor receptor variant III (EGFRvIII) has been investigated as a therapeutic target for chimeric antigen receptor (CAR) T cell therapy in glioblastoma. Earlier research demonstrated that phenotypic and genotypic characteristics in T cells and CAR T product predicted therapeutic success in hematologic malignancies, to date no determinants for clinical response in solid tumors have been identified. We analyzed apheresis and infusion products from the first-in-human trial of EGFRvIII-directed CAR T for recurrent glioblastoma (NCT02209376) by flow cytometry. Clinical response was quantified via engraftment in peripheral circulation and progression-free survival (PFS), as determined by the time from CAR T infusion to first radiographic evidence of progression. The CD4+CAR T cell population in patient infusion products demonstrated PD1 expression which positively correlated with AUC engraftment and PFS. On immune checkpoint inhibitor analysis, CTLA-4, TIM3, and LAG3 did not exhibit significant associations with engraftment or PFS. The frequencies of PD1+GZMB+ and PD1+HLA-DR+ CAR T cells in the CD4+ infusion products were directly proportional to AUC and PFS. No significant associations were observed within the apheresis products. In summary, PD1 in CAR T infusion products predicted peripheral engraftment and PFS in recurrent glioblastoma.

Keywords: CAR T cells; GBM; PD-1; glioblastoma; immunotherapy.

Conflict of interest statement

DO’R receives laboratory support from Tmunity Therapeutics. DO’R and ZB are on patents relating to CAR T cell therapy for GBM. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Tang, Tian, Yoder, Xu, Kulikovskaya, Gupta, Melenhorst, Lacey, O’Rourke and Binder.

Figures

Figure 1
Figure 1
Summary of Clinical Outcomes for EGFRvIII-Directed CAR T Clinical Trial and Flow Cytometric Analysis of Patient Plasma Products. Summary of clinical outcomes for clinical trial on EGFRvIII-directed CAR T for recurrent glioblastoma (NCT02209376). Patient 209 was ultimately not included for analysis due to having an outlier PFS value. (A) Plot of days following CAR T infusion and engraftment in peripheral blood. Following earlier studies, peripheral engraftment was quantified as log10copies/μg of genomic DNA. (B) PFS plotted as Kaplan-Meier estimator for all subjects. (C) Summary values of patient characteristics, performance of resection, total AUC, 30-day AUC, and peak engraftment for all subjects. * Patient 209 was excluded due to having an outlier progression-free survival (p<0.001 on Grubbs’ test). (D) Comparison of PD1 expression for CD3+CD4+CAR+ cells for transfusion products from all nine recurrent GBM patients receiving EGFRvIII-directed CAR T. Mean fluorescence intensity (MFI) is quantified on the table to the right. (E) Correlation of PD1 MFI and peak CAR T engraftment levels.
Figure 2
Figure 2
PD1 Correlations for CD4+CAR+ Cells in Patient Transduction Products. Correlation of PD1 expression with clinical outcomes for CD4+CAR+ cells in patient transduction products. (A) Association between PD1 expression and total AUC. (B) Association between PD1 expression and PFS. Error bars shown are 95% confidence intervals.
Figure 3
Figure 3
ICI Correlations for CD4+CAR+ Cells in Patient Transduction Products. Correlation of ICI expression (CTLA4, TIM3, LAG3) with clinical outcomes for CD4+CAR+ cells in patient transduction products. (A) Association between ICI expression and total AUC. (B) Association between ICI expression and PFS. (C) Correlation of PD1-ICI co-expression (PD1+CTLA4+, PD1+TIM3+, and PD1+LAG3+) and AUC. (D) Correlation of PD1-ICI co-expression and PFS. Error bars shown are 95% confidence intervals.
Figure 4
Figure 4
PD1 Correlations for CD4+CAR- Cells in Patient Transduction Products. Correlation of PD1 expression with clinical outcomes for CD4+CAR- cells in patient transduction products. (A) Association between PD1 expression and total AUC. (B) Association between PD1 expression and PFS. Error bars shown are 95% confidence intervals.
Figure 5
Figure 5
Activation Marker Correlations for CD4+CAR+ Cells in Patient Transduction Products. Correlation of activation marker expression (GRZB, HLA-DR) with clinical outcomes for CD4+CAR+ cells in patient transduction products. (A) Association between activation marker expression and total AUC. (B) Association between activation marker expression and PFS. (C) Correlation of PD1-activation marker co-expression (PD1+GRZB+ and PD1+HLA-DR+) and AUC. (D) Correlation of PD1-activation marker co-expression and PFS. Error bars shown are 95% confidence intervals.

References

    1. Koshy M, Villano JL, Dolecek TA, Howard A, Mahmood U, Chmura SJ, et al. . Improved Survival Time Trends for Glioblastoma Using the SEER 17 Population-Based Registries. J Neurooncol (2012) 107(1):207–12. doi: 10.1007/s11060-011-0738-7
    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. . Radiotherapy Plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N Engl J Med (2005) 352(10):987–96. doi: 10.1056/NEJMoa043330
    1. Patil CG, Yi A, Elramsisy A, Hu J, Mukherjee D, Irvin DK, et al. . Prognosis of Patients With Multifocal Glioblastoma: A Case-Control Study. J Neurosurg (2012) 117(4):705–11. doi: 10.3171/2012.7.JNS12147
    1. Rivera AL, Pelloski CE, Gilbert MR, Colman H, de la Cruz C, Sulman EP, et al. . MGMT Promoter Methylation is Predictive of Response to Radiotherapy and Prognostic in the Absence of Adjuvant Alkylating Chemotherapy for Glioblastoma. Neuro Oncol (2010) 12(2):116–21. doi: 10.1093/neuonc/nop020
    1. O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, et al. . A Single Dose of Peripherally Infused EGFRvIII-Directed CAR T Cells Mediates Antigen Loss and Induces Adaptive Resistance in Patients With Recurrent Glioblastoma. Sci Transl Med (2017) 9(399):eaaa0984. doi: 10.1126/scitranslmed.aaa0984
    1. Goff SL, Morgan RA, Yang JC, Sherry RM, Robbins PF, Restifo NP, et al. . Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-Transduced T Cells Targeting EGFRvIII in Patients With Glioblastoma. J Immunother (2019) 42(4):126–35. doi: 10.1097/CJI.0000000000000260
    1. Zhang C, Burger MC, Jennewein L, Genssler S, Schonfeld K, Zeiner P, et al. . ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J Natl Cancer Inst (2016) 108(5):djv375. doi: 10.1093/jnci/djv375
    1. Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. . Regression of Glioblastoma After Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med (2016) 375(26):2561–9. doi: 10.1056/NEJMoa1610497
    1. Li G, Wong AJ. EGF Receptor Variant III as a Target Antigen for Tumor Immunotherapy. Expert Rev Vaccines (2008) 7(7):977–85. doi: 10.1586/14760584.7.7.977
    1. Padfield E, Ellis HP, Kurian KM. Current Therapeutic Advances Targeting EGFR and EGFRvIII in Glioblastoma. Front Oncol (2015) 5:5. doi: 10.3389/fonc.2015.00005
    1. Chistiakov DA, Chekhonin IV, Chekhonin VP. The EGFR Variant III Mutant as a Target for Immunotherapy of Glioblastoma Multiforme. Eur J Pharmacol (2017) 810:70–82. doi: 10.1016/j.ejphar.2017.05.064
    1. Hamblett KJ, Kozlosky CJ, Siu S, Chang WS, Liu H, Foltz IN, et al. . AMG 595, an Anti-EGFRvIII Antibody-Drug Conjugate, Induces Potent Antitumor Activity Against EGFRvIII-Expressing Glioblastoma. Mol Cancer Ther (2015) 14(7):1614–24. doi: 10.1158/1535-7163.MCT-14-1078
    1. Schuster J, Lai RK, Recht LD, Reardon DA, Paleologos NA, Groves MD, et al. . Multicenter Trial of Rindopepimut (CDX-110) in Newly Diagnosed Glioblastoma: The ACT III Study. Neuro Oncol (2015) 17(6):854–61. doi: 10.1093/neuonc/nou348
    1. Durgin JS, Henderson F, Jr., Nasrallah MP, Mohan S, Wang S, Lacey SF, et al. . Case Report: Prolonged Survival Following EGFRvIII CAR T Cell Treatment for Recurrent Glioblastoma. Front Oncol (2021) 11:669071. doi: 10.3389/fonc.2021.669071
    1. Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, et al. . Determinants of Response and Resistance to CD19 Chimeric Antigen Receptor (CAR) T Cell Therapy of Chronic Lymphocytic Leukemia. Nat Med (2018) 24(5):563–71. doi: 10.1038/s41591-018-0010-1
    1. Garfall AL, Dancy EK, Cohen AD, Hwang WT, Fraietta JA, Davis MM, et al. . T-Cell Phenotypes Associated With Effective CAR T-Cell Therapy in Postinduction vs Relapsed Multiple Myeloma. Blood Adv (2019) 3(19):2812–5. doi: 10.1182/bloodadvances.2019000600
    1. Chongsathidkiet P, Jackson C, Koyama S, Loebel F, Cui X, Farber SH, et al. . Sequestration of T Cells in Bone Marrow in the Setting of Glioblastoma and Other Intracranial Tumors. Nat Med (2018) 24(9):1459–68. doi: 10.1038/s41591-018-0135-2
    1. Woroniecka KI, Rhodin KE, Chongsathidkiet P, Keith KA. Fecci Pe T-Cell Dysfunction in Glioblastoma: Applying a New Framework. Clin Cancer Res (2018) 24(16):3792–802. doi: 10.1158/1078-0432.CCR-18-0047
    1. Ohno M, Ohkuri T, Kosaka A, Tanahashi K, June CH, Natsume A, et al. . Expression of miR-17-92 Enhances Anti-Tumor Activity of T-Cells Transduced With the Anti-EGFRvIII Chimeric Antigen Receptor in Mice Bearing Human GBM Xenografts. J Immunother Cancer (2013) 1:21. doi: 10.1186/2051-1426-1-21
    1. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. . T Cells With Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients With Advanced Leukemia. Sci Transl Med (2011) 3(95):95ra73. doi: 10.1126/scitranslmed.3002842
    1. Levine BL, Miskin J, Wonnacott K, Keir C. Global Manufacturing of CAR T Cell Therapy. Mol Ther Methods Clin Dev (2017) 4:92–101. doi: 10.1016/j.omtm.2016.12.006
    1. Adusumilli PS, Zauderer MG, Riviere I, Solomon SB, Rusch VW, O'Cearbhaill RE, et al. . A Phase I Trial of Regional Mesothelin-Targeted CAR T-Cell Therapy in Patients With Malignant Pleural Disease, in Combination With the Anti-PD-1 Agent Pembrolizumab. Cancer Discovery (2021) 11(11):2748–63. doi: 10.1158/-21-0407
    1. Cao Y, Lu W, Sun R, Jin X, Cheng L, He X, et al. . Anti-CD19 Chimeric Antigen Receptor T Cells in Combination With Nivolumab Are Safe and Effective Against Relapsed/Refractory B-Cell Non-Hodgkin Lymphoma. Front Oncol (2019) 9:767. doi: 10.3389/fonc.2019.00767
    1. Chong EA, Melenhorst JJ, Lacey SF, Ambrose DE, Gonzalez V, Levine BL, et al. . PD-1 Blockade Modulates Chimeric Antigen Receptor (CAR)-Modified T Cells: Refueling the CAR. Blood (2017) 129(8):1039–41. doi: 10.1182/blood-2016-09-738245
    1. Gargett T, Yu W, Dotti G, Yvon ES, Christo SN, Hayball JD, et al. . GD2-Specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter But Can Be Protected From Activation-Induced Cell Death by PD-1 Blockade. Mol Ther (2016) 24(6):1135–49. doi: 10.1038/mt.2016.63
    1. Li AM, Hucks GE, Dinofia AM, Seif AE, Teachey DT, Baniewicz D, et al. . Checkpoint Inhibitors Augment CD19-Directed Chimeric Antigen Receptor (CAR) T Cell Therapy in Relapsed B-Cell Acute Lymphoblastic Leukemia. Blood (2018) 132:556. doi: 10.1182/blood-2018-99-112572
    1. Serganova I, Moroz E, Cohen I, Moroz M, Mane M, Zurita J, et al. . Enhancement of PSMA-Directed CAR Adoptive Immunotherapy by PD-1/PD-L1 Blockade. Mol Ther Oncolytics (2017) 4:41–54. doi: 10.1016/j.omto.2016.11.005
    1. Song W, Zhang M. Use of CAR-T Cell Therapy, PD-1 Blockade, and Their Combination for the Treatment of Hematological Malignancies. Clin Immunol (2020) 214:108382. doi: 10.1016/j.clim.2020.108382
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. . Tumor Antigen-Specific CD8 T Cells Infiltrating the Tumor Express High Levels of PD-1 and Are Functionally Impaired. Blood (2009) 114(8):1537–44. doi: 10.1182/blood-2008-12-195792
    1. Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, et al. . Upregulation of Tim-3 and PD-1 Expression Is Associated With Tumor Antigen-Specific CD8+ T Cell Dysfunction in Melanoma Patients. J Exp Med (2010) 207(10):2175–86. doi: 10.1084/jem.20100637
    1. Garzon-Muvdi T, Theodros D, Luksik AS, Maxwell R, Kim E, Jackson CM, et al. . Dendritic Cell Activation Enhances Anti-PD-1 Mediated Immunotherapy Against Glioblastoma. Oncotarget (2018) 9(29):20681–97. doi: 10.18632/oncotarget.25061
    1. Cherkassky L, Morello A, Villena-Vargas J, Feng Y, Dimitrov DS, Jones DR, et al. . Human CAR T Cells With Cell-Intrinsic PD-1 Checkpoint Blockade Resist Tumor-Mediated Inhibition. J Clin Invest (2016) 126(8):3130–44. doi: 10.1172/JCI83092
    1. Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, Lim WA, et al. . CRISPR/Cas9-Mediated PD-1 Disruption Enhances Anti-Tumor Efficacy of Human Chimeric Antigen Receptor T Cells. Sci Rep (2017) 7(1):737. doi: 10.1038/s41598-017-00462-8
    1. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM, et al. . Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors by Gene-Modified T Cells. Clin Cancer Res (2013) 19(20):5636–46. doi: 10.1158/1078-0432.CCR-13-0458
    1. DiDomenico J, Lamano JB, Oyon D, Li Y, Veliceasa D, Kaur G, et al. . The Immune Checkpoint Protein PD-L1 Induces and Maintains Regulatory T Cells in Glioblastoma. Oncoimmunology (2018) 7(7):e1448329. doi: 10.1080/2162402X.2018.1448329
    1. Kelly WJ, Giles AJ, Gilbert M. T Lymphocyte-Targeted Immune Checkpoint Modulation in Glioma. J Immunother Cancer (2020) 8(1):e000379. doi: 10.1136/jitc-2019-000379
    1. Finney OC, Brakke HM, Rawlings-Rhea S, Hicks R, Doolittle D, Lopez M, et al. . CD19 CAR T Cell Product and Disease Attributes Predict Leukemia Remission Durability. J Clin Invest (2019) 129(5):2123–32. doi: 10.1172/JCI125423
    1. Hong JJ, Amancha PK, Rogers K, Ansari AA, Villinger F. Re-Evaluation of PD-1 Expression by T Cells as a Marker for Immune Exhaustion During SIV Infection. PloS One (2013) 8(3):e60186. doi: 10.1371/journal.pone.0060186
    1. Kaiser AD, Schuster K, Gadiot J, Borkner L, Daebritz H, Schmitt C, et al. . Reduced Tumor-Antigen Density Leads to PD-1/PD-L1-Mediated Impairment of Partially Exhausted CD8(+) T Cells. Eur J Immunol (2012) 42(3):662–71. doi: 10.1002/eji.201141931
    1. Kinter AL, Godbout EJ, McNally JP, Sereti I, Roby GA, O'Shea MA, et al. . The Common Gamma-Chain Cytokines IL-2, IL-7, IL-15, and IL-21 Induce the Expression of Programmed Death-1 and Its Ligands. J Immunol (2008) 181(10):6738–46. doi: 10.4049/jimmunol.181.10.6738
    1. Wei J, Luo C, Wang Y, Guo Y, Dai H, Tong C, et al. . PD-1 Silencing Impairs the Anti-Tumor Function of Chimeric Antigen Receptor Modified T Cells by Inhibiting Proliferation Activity. J Immunother Cancer (2019) 7(1):209. doi: 10.1186/s40425-019-0685-y
    1. Wherry EJ, Kurachi M. Molecular and Cellular Insights Into T Cell Exhaustion. Nat Rev Immunol (2015) 15(8):486–99. doi: 10.1038/nri3862
    1. Davidson TB, Lee A, Hsu M, Sedighim S, Orpilla J, Treger J, et al. . Expression of PD-1 by T Cells in Malignant Glioma Patients Reflects Exhaustion and Activation. Clin Cancer Res (2019) 25(6):1913–22. doi: 10.1158/1078-0432.CCR-18-1176
    1. Gros A, Robbins PF, Yao X, Li YF, Turcotte S, Tran E, et al. . PD-1 Identifies the Patient-Specific CD8(+) Tumor-Reactive Repertoire Infiltrating Human Tumors. J Clin Invest (2014) 124(5):2246–59. doi: 10.1172/JCI73639
    1. Gustafson MP, DiCostanzo AC, Wheatley CM, Kim CH, Bornschlegl S, Gastineau DA, et al. . A Systems Biology Approach to Investigating the Influence of Exercise and Fitness on the Composition of Leukocytes in Peripheral Blood. J Immunother Cancer (2017) 5:30. doi: 10.1186/s40425-017-0231-8
    1. Chen X, Li X, Liu Y, Zhang Z, Zhang X, Huang J, et al. . A Phase I Clinical Trial of Chimeric Antigen Receptor-Modified T Cells in Patients With Relapsed and Refractory Lymphoma. Immunotherapy (2020) 12(10):681–96. doi: 10.2217/imt-2020-0022
    1. Lee YH, Lee HJ, Kim HC, Lee Y, Nam SK, Hupperetz C, et al. . PD-1 and TIGIT Downregulation Distinctly Affect the Effector and Early Memory Phenotypes of CD19-Targeting CAR T Cells. Mol Ther (2021) 30(2):579–92. doi: 10.1016/j.ymthe.2021.10.004
    1. Deng Q, Han G, Puebla-Osorio N, Ma MCJ, Strati P, Chasen B, et al. . Characteristics of Anti-CD19 CAR T Cell Infusion Products Associated With Efficacy and Toxicity in Patients With Large B Cell Lymphomas. Nat Med (2020) 26(12):1878–87. doi: 10.1038/s41591-020-1061-7
    1. Locke FL, Rossi JM, Neelapu SS, Jacobson CA, Miklos DB, Ghobadi A, et al. . Tumor Burden, Inflammation, and Product Attributes Determine Outcomes of Axicabtagene Ciloleucel in Large B-Cell Lymphoma. Blood Adv (2020) 4(19):4898–911. doi: 10.1182/bloodadvances.2020002394
    1. Bloch O, Crane CA, Kaur R, Safaee M, Rutkowski MJ, Parsa AT. Gliomas Promote Immunosuppression Through Induction of B7-H1 Expression in Tumor-Associated Macrophages. Clin Cancer Res (2013) 19(12):3165–75. doi: 10.1158/1078-0432.CCR-12-3314
    1. Gargett T, Truong N, Ebert LM, Yu W, Brown MP. Optimization of Manufacturing Conditions for Chimeric Antigen Receptor T Cells to Favor Cells With a Central Memory Phenotype. Cytotherapy (2019) 21(6):593–602. doi: 10.1016/j.jcyt.2019.03.003
    1. Gardner RA, Finney O, Annesley C, Brakke H, Summers C, Leger K, et al. . Intent-To-Treat Leukemia Remission by CD19 CAR T Cells of Defined Formulation and Dose in Children and Young Adults. Blood (2017) 129(25):3322–31. doi: 10.1182/blood-2017-02-769208
    1. Das RK, Vernau L, Grupp SA, Barrett DM. Naive T-Cell Deficits at Diagnosis and After Chemotherapy Impair Cell Therapy Potential in Pediatric Cancers. Cancer Discov (2019) 9(4):492–9. doi: 10.1158/-18-1314

Source: PubMed

3
Se inscrever